Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 348
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
J Cardiovasc Electrophysiol ; 35(5): 895-905, 2024 May.
Article in English | MEDLINE | ID: mdl-38433304

ABSTRACT

INTRODUCTION: Cardiac contractility modulation (CCM) is a medical device-based therapy delivering non-excitatory electrical stimulations to the heart to enhance cardiac function in heart failure (HF) patients. The lack of human in vitro tools to assess CCM hinders our understanding of CCM mechanisms of action. Here, we introduce a novel chronic (i.e., 2-day) in vitro CCM assay to evaluate the effects of CCM in a human 3D microphysiological system consisting of engineered cardiac tissues (ECTs). METHODS: Cryopreserved human induced pluripotent stem cell-derived cardiomyocytes were used to generate 3D ECTs. The ECTs were cultured, incorporating human primary ventricular cardiac fibroblasts and a fibrin-based gel. Electrical stimulation was applied using two separate pulse generators for the CCM group and control group. Contractile properties and intracellular calcium were measured, and a cardiac gene quantitative PCR screen was conducted. RESULTS: Chronic CCM increased contraction amplitude and duration, enhanced intracellular calcium transient amplitude, and altered gene expression related to HF (i.e., natriuretic peptide B, NPPB) and excitation-contraction coupling (i.e., sodium-calcium exchanger, SLC8). CONCLUSION: These data represent the first study of chronic CCM in a 3D ECT model, providing a nonclinical tool to assess the effects of cardiac electrophysiology medical device signals complementing in vivo animal studies. The methodology established a standardized 3D ECT-based in vitro testbed for chronic CCM, allowing evaluation of physiological and molecular effects on human cardiac tissues.


Subject(s)
Electrophysiologic Techniques, Cardiac , Myocardial Contraction , Myocytes, Cardiac , Myocardial Contraction/genetics , Myocardial Contraction/physiology , Tissue Engineering , Humans , Myocytes, Cardiac/physiology , Pluripotent Stem Cells/physiology , Gene Expression Profiling
3.
J Biomed Opt ; 28(1): 016001, 2023 01.
Article in English | MEDLINE | ID: mdl-36636698

ABSTRACT

Significance: All-optical cardiac electrophysiology enables the visualization and control of key parameters relevant to the detection of cardiac arrhythmias. Mapping such responses in human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) is of great interest for cardiotoxicity and personalized medicine applications. Aim: We introduce and validate a very low-cost compact mapping system for macroscopic all-optical electrophysiology in layers of hiPSC-CMs. Approach: The system uses oblique transillumination, low-cost cameras, light-emitting diodes, and off-the-shelf components (total < $ 15 , 000 ) to capture voltage, calcium, and mechanical waves under electrical or optical stimulation. Results: Our results corroborate the equivalency of electrical and optogenetic stimulation of hiPSC-CMs, and V m - [ Ca 2 + ] i similarity in conduction under pacing. Green-excitable optical sensors are combinable with blue optogenetic actuators (chanelrhodopsin2) only under very low green light ( < 0.05 mW / mm 2 ). Measurements in warmer culture medium yield larger spread of action potential duration and higher conduction velocities compared to Tyrode's solution at room temperature. Conclusions: As multiple optical sensors and actuators are combined, our results can help handle the "spectral congestion" and avoid parameter distortion. We illustrate the utility of the system for uncovering the action of cellular uncoupling agents and show extensibility to an epi-illumination mode for future imaging of thicker native or engineered tissues.


Subject(s)
Electrophysiologic Techniques, Cardiac , Induced Pluripotent Stem Cells , Humans , Myocytes, Cardiac/physiology , Arrhythmias, Cardiac , Action Potentials
4.
Am J Physiol Heart Circ Physiol ; 324(1): H141-H154, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36487188

ABSTRACT

Highlighting the importance of sex as a biological variable, we recently reported sex differences in guinea pig in vivo electrocardiogram (ECG) measurements. However, substantial inconsistencies exist in this animal model, with conflicting reports of sex-specific differences in cardiac electrophysiology observed in vivo and in vitro. Herein, we evaluated whether sexual dimorphism persists in ex vivo preparations, using an isolated intact heart preparation. Pseudo-ECG recordings were collected in conjunction with dual optical mapping of transmembrane voltage and intracellular calcium from Langendorff-perfused hearts. In contrast to our in vivo results, we did not observe sex-specific differences in ECG parameters collected from isolated hearts. Furthermore, we observed significant age-specific differences in action potential duration (APD) and Ca2+ transient duration (CaD) during both normal sinus rhythm (NSR) and in response to dynamic pacing but only a modest sex-specific difference in CaD30. Similarly, the alternans fluctuation coefficient, conduction velocity during sinus rhythm or in response to pacing, and electrophysiology parameters (atrioventricular nodal effective refractory period, Wenckebach cycle length) were comparable between males and females. Results of our study suggest that the observed sex-specific differences in in vivo ECG parameters from guinea pigs are diminished in ex vivo isolated heart preparations, although age-specific patterns are prevalent. To assess sex as a biological variable in cardiac electrophysiology, a comprehensive approach may be necessary using both in vitro measurements from cardiomyocyte or intact heart preparations with secondary follow-up in vivo studies.NEW & NOTEWORTHY We evaluated whether the guinea pig heart has intrinsic sex-specific differences in cardiac electrophysiology. Although we observed sex-specific differences in in vivo ECGs, these differences did not persist ex vivo. Using a whole heart model, we observed similar APD, CaD, conduction velocity, and alternans susceptibility in males and females. We conclude that sex-specific differences in guinea pig cardiac electrophysiology are likely influenced by the in vivo environment and less dependent on the intrinsic electrical properties of the heart.


Subject(s)
Electrophysiologic Techniques, Cardiac , Heart Conduction System , Guinea Pigs , Female , Animals , Male , Heart/physiology , Electrocardiography , Myocytes, Cardiac/physiology , Arrhythmias, Cardiac , Action Potentials
5.
Methods Mol Biol ; 2485: 213-225, 2022.
Article in English | MEDLINE | ID: mdl-35618908

ABSTRACT

Different engineered heart muscle formats have been developed for applications in disease modeling, drug screening, and heart repair. The advantage of 3D engineered versus 2D monolayer and 3D aggregate cardiomyocyte cultures is a clearly advanced degree of maturation, which in many aspects resembles the postnatal rather than the embryonic or fetal heart, in the most advanced 3D culture formats. According to the desired in vitro (disease modeling or drug screening) and in vivo (heart repair) application, scale and geometry of tissue engineered heart muscle must be adapted. In this updated methods paper, we report a simple and scalable (up and down) collagen-based protocol for the construction of Engineered Human Myocardium (EHM) under defined, serum-free conditions.


Subject(s)
Cardiac Surgical Procedures , Myocardium , Drug Evaluation, Preclinical , Humans , Myocytes, Cardiac/physiology , Tissue Engineering/methods
6.
J Physiol ; 600(9): 2037-2048, 2022 05.
Article in English | MEDLINE | ID: mdl-35301719

ABSTRACT

Daily variations in cardiac electrophysiology and the incidence for different types of arrhythmias reflect ≈24 h changes in the environment, behaviour and internal circadian rhythms. This article focuses on studies that use animal models to separate the impact that circadian rhythms, as well as changes in the environment and behaviour, have on 24 h rhythms in heart rate and ventricular repolarization. Circadian rhythms are initiated at the cellular level by circadian clocks, transcription-translation feedback loops that cycle with a periodicity of 24 h. Several studies now show that the circadian clock in cardiomyocytes regulates the expression of cardiac ion channels by multiple mechanisms; underlies time-of-day changes in sinoatrial node excitability/intrinsic heart rate; and limits the duration of the ventricular action potential waveform. However, the 24 h rhythms in heart rate and ventricular repolarization are primarily driven by autonomic signalling. A functional role for the cardiomyocyte circadian clock appears to buffer the heart against perturbations. For example, the cardiomyocyte circadian clock limits QT-interval prolongation (especially at slower heart rates), and it may facilitate the realignment of the 24 h rhythm in heart rate to abrupt changes in the light cycle. Additional studies show that modifying rhythmic behaviours (including feeding behaviour) can dramatically impact the 24 h rhythms in heart rate and ventricular repolarization. If these mechanisms are conserved, these studies suggest that targeting endogenous circadian mechanisms in the heart, as well as modifying the timing of certain rhythmic behaviours, could emerge as therapeutic strategies to support heart function against perturbations and regulate 24 h rhythms in cardiac electrophysiology.


Subject(s)
Circadian Clocks , Animals , Circadian Clocks/physiology , Circadian Rhythm/physiology , Electrophysiologic Techniques, Cardiac , Ion Channels/metabolism , Myocytes, Cardiac/physiology
7.
Toxicol Appl Pharmacol ; 438: 115914, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35150662

ABSTRACT

The goal of the CiPA initiative (Comprehensive in vitro Proarrhythmia Assay) was to assess a more accurate prediction of new drug candidate proarrhythmic severe liabilities such as torsades de pointes, for example. This new CiPA paradigm was partly based on in silico reconstruction of human ventricular cardiomyocyte action potential useful to identify repolarization abnormalities such early afterdepolarization (EAD), for example. Using the ToR-ORd algorithm (Tomek-Rodriguez-O'Hara-Rudy dynamic model), the aim of the present work was (i) to identify intracellular parameters leading to EAD occurrence under healthy and hypertrophic cardiomyopathy (HCM) conditions and (ii) to evaluate the prediction accuracy of compound torsadogenic risk based on EAD occurrence using a large set of 109 torsadogenic and non-torsadogenic compounds under both experimental conditions. In silico results highlighted the crucial involvement of Ca++ handling in the ventricular cardiomyocyte intracellular subspace compartment for the initiation of EAD, demonstrated by a higher amplitude of Ca++ release from junctional sarcoplasmic reticulum to subspace compartments (Jrel) measured at EAD take-off voltage in the presence vs. the absence of EAD initiated either by high IKr inhibition or by high enough concentration of a torsadogenic compound under both experimental conditions. Under healthy or HCM conditions, the prediction accuracy of the torsadogenic risk of compound based on EAD occurrence was observed to be 61 or 92%, respectively. This high accuracy under HCM conditions was discussed regarding its usefulness for cardiac safety pharmacology at least at early drug screening/preclinical stage of the drug development process.


Subject(s)
Action Potentials/physiology , Cardiomyopathy, Hypertrophic/drug therapy , Cardiovascular Agents/pharmacology , Myocytes, Cardiac/drug effects , Torsades de Pointes/drug therapy , Algorithms , Calcium/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Computer Simulation , Drug Evaluation, Preclinical/methods , Electrocardiography/drug effects , Humans , Myocytes, Cardiac/physiology , Risk Assessment , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Torsades de Pointes/physiopathology
8.
Int J Immunopathol Pharmacol ; 36: 20587384211051993, 2022.
Article in English | MEDLINE | ID: mdl-34986670

ABSTRACT

OBJECTIVES: Alantolactone (AL) is a compound extracted from the roots of Inula Racemosa that has shown beneficial effects in cardiovascular disease. However, the cardioprotective mechanism of AL against hypoxic/ischemic (H/I) injury is still unclear. This research aimed to determine AL's ability to protect the heart against isoproterenol (ISO)-induced MI injury in vivo and cobalt chloride (CoCl2) induced H/I injury in vitro. METHODS: Electrocardiography (ECG), lactate dehydrogenase (LDH), creatine kinase (CK), and cardiac troponin I (cTnI) assays in addition to histological analysis of the myocardium were used to investigate the effects of AL in vivo. Influences of AL on L-type Ca2+ current (ICa-L) in isolated rat myocytes were observed by the patch-clamp technique. Furthermore, cell viability, apoptosis, oxidative stress injury, mitochondrial membrane potential, and intracellular Ca2+ concentration were examined in vitro. RESULTS: The results indicated that AL treatment ameliorated the morphological and ECG changes associated with MI, and decreased levels of LDH, CK, and cTnI. Furthermore, pretreatment with AL elevated antioxidant enzyme activity and suppressed ROS production. AL prevented H/I-induced apoptosis, mitochondria damage, and calcium overload while reducing ICa-L in a concentration and time dependent fashion. The 50% inhibiting concentration (IC50) and maximal inhibitory effect (Emax) of AL were 17.29 µmol/L and 57.73 ± 1.05%, respectively. CONCLUSION: AL attenuated MI-related injury by reducing oxidative stress, apoptosis, calcium overload, and mitochondria damage. These cardioprotective effects may be related to the direct inhibition of ICa-L.


Subject(s)
Cardiotonic Agents/therapeutic use , Lactones/therapeutic use , Myocardial Ischemia/drug therapy , Sesquiterpenes, Eudesmane/therapeutic use , Animals , Apoptosis/drug effects , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cell Line , Cobalt/toxicity , Heart Rate/drug effects , Interleukin-6/metabolism , Isoproterenol , Lactones/pharmacology , Male , Myocardial Ischemia/chemically induced , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sesquiterpenes, Eudesmane/pharmacology , Tumor Necrosis Factor-alpha/metabolism
9.
Transl Res ; 243: 60-77, 2022 05.
Article in English | MEDLINE | ID: mdl-35077866

ABSTRACT

Biomarkers of mineral bone disorders (MBD) including phosphorus, fibroblast growth factor (FGF)-23 and Klotho are strongly altered in patients with acute kidney injury (AKI) who have high cardiac outcomes and mortality rates. However, the crosslink between MBD and cardiac damage after an AKI episode still remains unclear. We tested MBD and cardiac biomarkers in an experimental AKI model after 24 or 72 hours of folic acid injection and we analyzed structural cardiac remodeling, intracellular calcium (Ca2+) dynamics in cardiomyocytes and cardiac rhythm. AKI mice presented high levels of FGF-23, phosphorus and cardiac troponin T and exhibited a cardiac hypertrophy phenotype accompanied by an increase in systolic Ca2+ release 24 hours after AKI. Ca2+ transients and contractile dysfunction were reduced 72 hours after AKI while diastolic sarcoplasmic reticulum Ca2+ leak, pro-arrhythmogenic Ca2+ events and ventricular arrhythmias were increased. These cardiac events were linked to the activation of the calcium/calmodulin-dependent kinase II pathway through the increased phosphorylation of ryanodine receptors and phospholamban specific sites after AKI. Cardiac hypertrophy and the altered intracellular Ca2+ dynamics were prevented in transgenic mice overexpressing Klotho after AKI induction. In a translational retrospective longitudinal clinical study, we determined that combining FGF-23 and phosphorus with cardiac troponin T levels achieved a better prediction of mortality in AKI patients at hospital admission. Thus, monitoring MBD and cardiac damage biomarkers could be crucial to prevent mortality in AKI patients. In this setting, Klotho might be considered as a new cardioprotective therapeutic tool to prevent deleterious cardiac events in AKI conditions.


Subject(s)
Acute Kidney Injury , Calcium , Acute Kidney Injury/etiology , Animals , Arrhythmias, Cardiac , Biomarkers/metabolism , Calcium/metabolism , Cardiomegaly/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Minerals/metabolism , Myocytes, Cardiac/physiology , Phosphorus/metabolism , Retrospective Studies , Troponin T/metabolism
10.
Nutrients ; 13(11)2021 Oct 30.
Article in English | MEDLINE | ID: mdl-34836155

ABSTRACT

Ursolic acid (UA) is a natural pentacyclic triterpenoid found in a number of plants such as apples, thyme, oregano, hawthorn and others. Several in vitro and in vivo studies have presented its anti-inflammatory and anti-apoptotic properties. The inhibition of NF-κB-mediated inflammatory pathways and the increased scavenging of reactive oxygen species (ROS) in numerous ways seem to be the most beneficial effects of UA. In mice and rats, administration of UA appears to slow down the development of cardiovascular diseases (CVDs), especially atherosclerosis and cardiac fibrosis. Upregulation of endothelial-type nitric oxide synthase (eNOS) and cystathionine-λ-lyase (CSE) by UA may suggest its vasorelaxant property. Inhibition of metalloproteinases activity by UA may contribute to better outcomes in aneurysms management. UA influence on lipid and glucose metabolism remains inconsistent, and additional studies are essential to verify its efficacy. Furthermore, UA derivatives appear to have a beneficial impact on the cardiovascular system. This review aims to summarize recent findings on beneficial effects of UA that may make it a promising candidate for clinical trials for the management of CVDs.


Subject(s)
Cardiovascular Diseases/diet therapy , Dietary Supplements , Triterpenes/administration & dosage , Triterpenes/pharmacology , Animals , Atherosclerosis/diet therapy , Cardiovascular System/drug effects , Clinical Trials as Topic , Female , Humans , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Vasodilation/drug effects , Ursolic Acid
11.
J Cell Mol Med ; 25(24): 11257-11263, 2021 12.
Article in English | MEDLINE | ID: mdl-34761519

ABSTRACT

Cardiovascular disease is a leading cause of death and disability worldwide. Although genetically modified mouse models offer great potential for robust research in vivo, in vitro studies using isolated cardiomyocytes also provide an important approach for investigating the mechanisms underlying cardiovascular disease pathogenesis and drug actions. Currently, isolation of mouse adult cardiomyocytes often relies on aortic retrograde intubation under a stereoscopic microscope, which poses considerable technical barriers and requires extensive training. Although a simplified, Langendorff-free method has been used to isolate viable cardiomyocytes from the adult mouse heart, the system requires enzymatic digestions and continuous manual technical operation. This study established an optimized approach that allows isolation of adult mouse cardiomyocytes and epicardial activation mapping of mouse hearts using a Langendorff device. We used retrograde puncture through the abdominal aorta in vivo and enzymatic digestion on the Langendorff perfusion device to isolate adult mouse cardiomyocytes without using a microscope. The yields of isolated cardiomyocytes were amenable to patch clamp techniques. Furthermore, this approach allowed epicardial activation mapping. We used a novel, simplified method to isolate viable cardiomyocytes from adult mouse hearts and to map epicardial activation. This novel approach could be beneficial in more extensive research in the cardiac field.


Subject(s)
Cell Separation , Epicardial Mapping , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Action Potentials , Animals , Cell Culture Techniques , Cell Separation/methods , Drug Evaluation, Preclinical , Electrophysiologic Techniques, Cardiac , Epicardial Mapping/methods , Mice , Myocytes, Cardiac/drug effects , Patch-Clamp Techniques
12.
Food Funct ; 12(19): 9360-9371, 2021 Oct 04.
Article in English | MEDLINE | ID: mdl-34606545

ABSTRACT

Anhydrosafflor yellow B (AHSYB) is a major active water-soluble pigment in Safflower, but it has not received enough attention yet. In this study, high-speed counter-current chromatography (HSCCC) was used to prepare AHSYB from safflower. The parameters of the separation process were optimized by response surface methodology for the first time. The entropy weight method (EWM) was applied to calculate the information entropy and the weight of five indexes, and then figure out a comprehensive index of the HSCCC separation effect. Under the optimized separation conditions, a HSCCC apparatus speed of 850 rpm, a flow rate of 2 mL min-1 for the mobile phase and a separation temperature of 40 °C for AHSYB were achieved with a purity of 98%. Furthermore, AHSYB was found to have cardio-protective effects by inhibiting apoptosis via the mitochondrial-mediated pathway in oxygen-glucose deprivation/reoxygenation-induced H9c2 cells. This research provides good method guides for the rapid and efficient separation of active compounds from food-grade Chinese herb medicines.


Subject(s)
Apoptosis/drug effects , Cardiotonic Agents/isolation & purification , Cardiotonic Agents/pharmacology , Carthamus tinctorius/chemistry , Myocytes, Cardiac/drug effects , Pigments, Biological/isolation & purification , Pigments, Biological/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cardiotonic Agents/chemistry , Caspase 3/genetics , Caspase 3/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Countercurrent Distribution , Cytochromes c/genetics , Cytochromes c/metabolism , Down-Regulation , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Pigments, Biological/chemistry , Plant Extracts/chemistry , Rats , Reactive Oxygen Species
13.
Nat Commun ; 12(1): 5804, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608155

ABSTRACT

During the last decade, cardiac optogenetics has turned into an essential tool for investigating cardiac function in general and for assessing functional interactions between different myocardial cell types in particular. To advance exploitation of the unique research opportunities offered by this method, we develop a panoramic opto-electrical measurement and stimulation (POEMS) system for mouse hearts. The core of the experimental platform is composed of 294 optical fibers and 64 electrodes that form a cup which embraces the entire ventricular surface of mouse hearts and enables straightforward 'drop&go' experimentation. The flexible assignment of fibers and electrodes to recording or stimulation tasks permits a precise tailoring of experiments to the specific requirements of individual optogenetic constructs thereby avoiding spectral congestion. Validation experiments with hearts from transgenic animals expressing the optogenetic voltage reporters ASAP1 and ArcLight-Q239 demonstrate concordance of simultaneously recorded panoramic optical and electrical activation maps. The feasibility of single fiber optical stimulation is proven with hearts expressing the optogenetic voltage actuator ReaChR. Adaptation of the POEMS system to larger hearts and incorporation of additional sensors can be achieved by redesigning the system-core accordingly.


Subject(s)
Heart/physiology , Optogenetics/methods , Animals , Electrophysiologic Techniques, Cardiac , Heart Rate , Membrane Potentials , Mice , Mice, Transgenic , Myocytes, Cardiac/physiology , Optogenetics/instrumentation , Voltage-Sensitive Dye Imaging
14.
Adv Sci (Weinh) ; 8(21): e2100627, 2021 11.
Article in English | MEDLINE | ID: mdl-34486241

ABSTRACT

Optical stimulation technologies are gaining great consideration in cardiology, neuroscience studies, and drug discovery pathways by providing control over cell activity with high spatio-temporal resolution. However, this high precision requires manipulation of biological processes at genetic level concealing its development from broad scale application. Therefore, translating these technologies into tools for medical or pharmacological applications remains a challenge. Here, an all-optical nongenetic method for the modulation of electrogenic cells is introduced. It is demonstrated that plasmonic metamaterials can be used to elicit action potentials by converting near infrared laser pulses into stimulatory currents. The suggested approach allows for the stimulation of cardiomyocytes and neurons directly on commercial complementary metal-oxide semiconductor microelectrode arrays coupled with ultrafast pulsed laser, providing both stimulation and network-level recordings on the same device.


Subject(s)
Action Potentials/drug effects , Infrared Rays , Myocytes, Cardiac/physiology , Nanostructures/toxicity , Neurons/physiology , Action Potentials/radiation effects , Animals , Cell Line , Humans , Metals/chemistry , Mice , Microelectrodes , Myocytes, Cardiac/cytology , Nanostructures/chemistry , Neurons/cytology , Porosity , Rats , Semiconductors , Silicon Dioxide/chemistry
15.
Elife ; 102021 07 02.
Article in English | MEDLINE | ID: mdl-34212860

ABSTRACT

The development of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has been a critical in vitro advance in the study of patient-specific physiology, pathophysiology, and pharmacology. We designed a new deep learning multitask network approach intended to address the low throughput, high variability, and immature phenotype of the iPSC-CM platform. The rationale for combining translation and classification tasks is because the most likely application of the deep learning technology we describe here is to translate iPSC-CMs following application of a perturbation. The deep learning network was trained using simulated action potential (AP) data and applied to classify cells into the drug-free and drugged categories and to predict the impact of electrophysiological perturbation across the continuum of aging from the immature iPSC-CMs to the adult ventricular myocytes. The phase of the AP extremely sensitive to perturbation due to a steep rise of the membrane resistance was found to contain the key information required for successful network multitasking. We also demonstrated successful translation of both experimental and simulated iPSC-CM AP data validating our network by prediction of experimental drug-induced effects on adult cardiomyocyte APs by the latter.


Subject(s)
Algorithms , Deep Learning , Electrophysiologic Techniques, Cardiac , Myocytes, Cardiac/physiology , Action Potentials/physiology , Cell Differentiation/physiology , Computer Simulation , ERG1 Potassium Channel/genetics , ERG1 Potassium Channel/metabolism , Electrophysiological Phenomena/physiology , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/physiology , Models, Biological , Phenethylamines/pharmacology , Sulfonamides/pharmacology
16.
Int Immunopharmacol ; 98: 107841, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34153662

ABSTRACT

BACKGROUND: Ginsenoside Rg3 (Rg3), one of the most potent components extracted from the roots of the traditional Chinese herb Panax ginseng, has prominent roles in anti-tumor and anti-inflammation. However, the applications of Rg3 against myocardial hypertrophy are not fully revealed. METHODS: Transverse aortic constriction (TAC) was adopted to build the myocardial hypertrophy model in rats. The in vitro model of myocardial hypertrophy was induced by angiotensin II (Ang II) in the human cardiomyocyte cell line AC16 and HCM, which were then treated with different doses of Rg3. The levels of myocardial hypertrophy markers (ANP, BNP, and ß-MHC) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot (WB) was conducted to verify the expressions of myocardial fibrosis-associated proteins (MyHc, Collagen Ⅰ, and TGF-ß1) and oxidative stress (OS) proteins (HO-1 and Nrf2). The markers of fibrosis, hypertrophy, NLRP3 inflammasome and OS in cardiomyocytes were evaluated by qRT-PCR, western blot (WB), enzyme-linked immunosorbent assay (ELISA), and cellular immunofluorescence, respectively. Furthermore, pharmacological intervention on sirtuin-1 (SIRT1) was performed to clarify the function of SIRT1 in Rg3-mediated effects. RESULTS: Rg3 dose-dependently attenuated the Ang II-induced myocardial hypertrophy and fibrosis. What's more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. Mechanistically, Rg3 attenuated NF-κB activation and promoted SIRT1 expression. Inhibiting SIRT1 (by AGK2) mostly reversed Rg3-mediated effects against Ang II-induced myocardial hypertrophy and fibrosis. In the TAC rat model, administration of Rg3 mitigated myocardial hypertrophy and fibrosis through pressing overproduced inflammation and OS. CONCLUSION: Rg3 prevents Ang II-induced myocardial hypertrophy via inactivating NLRP3 inflammasome and oxidative stress by modulating the SIRT1/NF-κB pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Ginsenosides/therapeutic use , Hypertrophy/drug therapy , Inflammasomes/metabolism , Myocardium/pathology , Myocytes, Cardiac/physiology , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sirtuin 1/metabolism , Angiotensin II/metabolism , Animals , Aorta/surgery , Cells, Cultured , Disease Models, Animal , Fibrosis , Humans , Immunomodulation , Myocardium/metabolism , Oxidative Stress , Rats , Signal Transduction
17.
Aquat Toxicol ; 235: 105823, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33906022

ABSTRACT

Oil and gas exploration in the Arctic can result in the release of polycyclic aromatic hydrocarbons (PAHs) into relatively pristine environments. Following the recent spill of approximately 17 500 tonnes of diesel fuel in Norilsk, Russia, May 2020, our study focussed on the effects of phenanthrene, a low molecular weight PAH found in diesel and crude oil, on the isolated atrial and ventricular myocytes from the heart of the polar teleost, the Navaga cod (Eleginus nawaga). Acute exposure to phenanthrene in navaga cardiomyocytes caused significant action potential (AP) prolongation, confirming the proarrhythmic effects of this pollutant. We show AP prolongation was due to potent inhibition of the main repolarising current, IKr, with an IC50 value of ~2 µM. We also show a potent inhibitory effect (~55%) of 1 µM phenanthrene on the transient IKr currents that protects the heart from early-after-depolarizations and arrhythmias. These data, along with more minor effects on inward sodium (INa) (~17% inhibition at 10 µM) and calcium (ICa) (~17% inhibition at 30 µM) currents, and no effects on inward rectifier (IK1 and IKAch) currents, demonstrate the cardiotoxic effects exerted by phenanthrene on the atrium and ventricle of navaga cod. Moreover, we report the first data that we are aware of on the impact of phenanthrene on atrial myocyte function in any fish species.


Subject(s)
Gadiformes/physiology , Myocytes, Cardiac/drug effects , Phenanthrenes/toxicity , Water Pollutants, Chemical/toxicity , Action Potentials/drug effects , Animals , Arctic Regions , Fishes , Myocytes, Cardiac/physiology , Petroleum , Polycyclic Aromatic Hydrocarbons/toxicity , Sodium/pharmacology
18.
AAPS J ; 23(2): 44, 2021 03 14.
Article in English | MEDLINE | ID: mdl-33719006

ABSTRACT

Anthracyclines are a class of chemotherapy drugs that are highly effective for the treatment of human cancers, but their clinical use is limited by associated dose-dependent cardiotoxicity. The precise mechanisms by which individual anthracycline induces cardiotoxicity are not fully understood. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are emerging as a physiologically relevant model to assess drugs cardiotoxicity. Here, we describe an assay platform by coupling hiPSC-CMs and impedance measurement, which allows real-time monitoring of cardiomyocyte cellular index, beating amplitude, and beating rate. Using this approach, we have performed comparative studies on a panel of four anthracycline drugs (doxorubicin, epirubicin, idarubicin, and daunorubicin) which share a high degree of structural similarity but are associated with distinct cardiotoxicity profiles and maximum cumulative dose limits. Notably, results from our hiPSC-CMs impedance model (dose-dependent responses and EC50 values) agree well with the recommended clinical dose limits for these drugs. Using time-lapse imaging and RNAseq, we found that the differences in anthracycline cardiotoxicity are closely linked to extent of cardiomyocyte uptake and magnitude of activation/inhibition of several cellular pathways such as death receptor signaling, ROS production, and dysregulation of calcium signaling. The results provide molecular insights into anthracycline cardiac interactions and offer a novel assay system to more robustly assess potential cardiotoxicity during drug development.


Subject(s)
Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Myocytes, Cardiac/drug effects , Biological Assay/methods , Calcium Signaling/drug effects , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Electric Impedance , Humans , Induced Pluripotent Stem Cells/physiology , Intravital Microscopy/methods , Myocytes, Cardiac/physiology , Oxidative Stress/drug effects , RNA-Seq , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time-Lapse Imaging
19.
Pflugers Arch ; 473(7): 1087-1097, 2021 07.
Article in English | MEDLINE | ID: mdl-33686477

ABSTRACT

Heart diseases are prevalent worldwide and account for the highest mortality than any other illness. Although investment in drug discovery and development has increased, the amount of drug approvals has seen a progressive decline. Moreover, adverse side effects to the heart have become the most common reasons for preclinical project cessation, partly due to the lack of suitable humanized preclinical models. Human pluripotent stem cells (hPSCs) have emerged as a powerful non-animal platform to model heart disease, to screen for novel drugs, and to test drug cardiotoxicity in a high-throughput and cost-effective manner. Here, we review and discuss recent breakthroughs in the development of cardiovascular modeling and their current and future applications of hPSC-based drug discovery and testing.


Subject(s)
Cardiovascular Diseases/pathology , Pluripotent Stem Cells/physiology , Animals , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Humans , Myocytes, Cardiac/physiology
20.
Int J Mol Sci ; 22(5)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673574

ABSTRACT

Offspring born to diabetic or obese mothers have a higher lifetime risk of heart disease. Previously, we found that rat offspring exposed to late-gestational diabetes mellitus (LGDM) and maternal high-fat (HF) diet develop mitochondrial dysfunction, impaired cardiomyocyte bioenergetics, and cardiac dysfunction at birth and again during aging. Here, we compared echocardiography, cardiomyocyte bioenergetics, oxidative damage, and mitochondria-mediated cell death among control, pregestational diabetes mellitus (PGDM)-exposed, HF-diet-exposed, and combination-exposed newborn offspring. We hypothesized that PGDM exposure, similar to LGDM, causes mitochondrial dysfunction to play a central, pathogenic role in neonatal cardiomyopathy. We found that PGDM-exposed offspring, similar to LGDM-exposed offspring, have cardiac dysfunction at birth, but their isolated cardiomyocytes have seemingly less bioenergetics impairment. This finding was due to confounding by impaired viability related to poorer ATP generation, more lipid peroxidation, and faster apoptosis under metabolic stress. To mechanistically isolate and test the role of mitochondria, we transferred mitochondria from normal rat myocardium to control and exposed neonatal rat cardiomyocytes. As expected, transfer provides a respiratory boost to cardiomyocytes from all groups. They also reduce apoptosis in PGDM-exposed males, but not in females. Findings highlight sex-specific differences in mitochondria-mediated mechanisms of developmentally programmed heart disease and underscore potential caveats of therapeutic mitochondrial transfer.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetes, Gestational/physiopathology , Energy Metabolism , Heart Diseases/prevention & control , Mitochondria/transplantation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Animals , Diet, High-Fat/adverse effects , Female , Heart Diseases/etiology , Heart Diseases/pathology , Male , Maternal Nutritional Physiological Phenomena , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/prevention & control , Rats , Rats, Sprague-Dawley , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL